Author Archives: Tyler

Blood Pressure Cuffs versus LSJL

Does LSJL only increase intermedullary pressure or does it have extra anabolic effects?  The next few studies address what happens only if intermedullary pressure is increased.  It’s possible that LSJL’s effects are due solely to an increase in vascularization which makes it less promising for adult height growth although it would still be possible to induce adult height growth.  These studies are performed on rats much older than those used in LSJL but no impact on longitudinal bone growth was studied.  However, it still shows even in aged individuals developmental genes like BMP-2 and TGF-Beta can be stimulated.

Dynamic hydraulic fluid stimulation regulated intramedullary pressure

“Physical signals within the bone, i.e. generated from mechanical loading, have the potential to initiate skeletal adaptation. Strong evidence has pointed to bone fluid flow (BFF) as a media between an external load and the bone cells, in which altered velocity and pressure can ultimately initiate the mechanotransduction and the remodeling process within the bone. Load-induced BFF can be altered by factors such as intramedullary pressure (ImP) and/or bone matrix strain, mediating bone adaptation. BFF induced by ImP alone, with minimum bone strain, can initiate bone remodeling.  To apply ImP as a means for alteration of BFF, it was hypothesized that non-invasive dynamic hydraulic stimulation (DHS) can induce local ImP with minimal bone strain to potentially elicit osteogenic adaptive responses via bone–muscle coupling. The goal of this study was to evaluate the immediate effects on local and distant ImP and strain in response to a range of loading frequencies using DHS. Simultaneous femoral and tibial ImP and bone strain values were measured in three 15-month-old female Sprague Dawley rats during DHS loading on the tibia with frequencies of 1 Hz to 10 Hz. DHS showed noticeable effects on ImP induction in the stimulated tibia in a nonlinear fashion in response to DHS over the range of loading frequencies, where they peaked at 2 Hz. DHS at various loading frequencies generated minimal bone strain in the tibiae. Maximal bone strain measured at all loading frequencies was less than 8 με. No detectable induction of ImP or bone strain was observed in the femur. This study suggested that oscillatory DHS may regulate the local fluid dynamics with minimal mechanical strain in the bone, which serves critically in bone adaptation. These results clearly implied DHS’s potential as an effective, non-invasive intervention for osteopenia and osteoporosis treatments.”

“bone fluid flow (BFF) with altered velocity or pressure acts as a communication media between an external load and the bone cells, which then regulate bone remodeling. In converse, discontinuous BFF can initiate bone turnover and result in osteopenia”

“Induced ImP possibly triggers the transformation of the bone nutrient vasculature, leading to the ultimate alteration in blood supply to the bone.”

“The ImP reached the peak at 2 Hz.” at about 15mmHg.

Dynamic fluid flow stimulation on cortical bone and alterations of the gene expressions of osteogenic growth factors and transcription factors in a rat functional disuse model

“Dynamic hydraulic stimulation (DHS) [is] a loading modality to induce anabolic responses in bone. To further study the functional process of DHS regulated bone metabolism, the objective of this study was to evaluate the effects of DHS on cortical bone and its alterations on gene expressions of osteogenic growth factors and transcription factors as a function of time. Using a model system of 5-month-old hindlimb suspended (HLS) female Sprague–Dawley rats, DHS was applied to the right tibiae of the stimulated rats with a loading frequency of 2 Hz with 30 mmHg (p–p) dynamic pressure, 5 days/week, for a total of 28 days. Midshafts of the tibiae were analyzed using μCT and histology. Total RNA was analyzed using RT-PCR on selected osteogenic genes (RUNX2, β-catenin, osteopontin, VEGF, BMP2, IGF-1, and TGF-β) on 3-, 7-, 14- , and 21-day. Results showed increased Cort.Th and Ct.BV/TV as well as a time-dependent fashion of gradual changes in mRNA levels upon DHS. While DHS-driven fold changes of the mRNA levels remained low before Day-7, its fold changes started to elevate by Day-14 and then dropped by Day-21. This study further delineates the underlying molecular mechanism of DHS-derived mechanical signals, and its time-dependent optimization.”

BMP2, IGF-1 and Tgf-Beta are all pro chondrogenic.

“Bone formation progresses over time after the initiation of mechanical loading. Within 24–48 hr after mechanical loading starts, new osteoblasts lay on the bone surface and contribute to bone formation that is observed after 96 hr of loading. Bone formation follows a time-dependent manner, in which it increases between 5 and 12 days of loading and returns back to baseline levels after 6 weeks of loading. These data suggest that the whole cycle of bone formation, including osteoblast recruitment followed by matrix production, lasts for about 5 weeks before declining back to baseline levels”<-new osteoblasts implies cell differentiation which could mean new chondrocytes as well.

Dynamic Hydraulic stimulation increased the periosteal surface.

At seven days interestingly the levels of the pro-chondrogenic genes IGF-1, BMP2, and TGF-Beta were low.  At 14 days they were all increased relative to control.

“HS may compress the veins within the skeletal muscle and increase the vasculature pressure gradient that promotes capillary blood flow in bone. Increase of capillary filtration may further increase ImP and induce BFF that ultimately promote bone regeneration”

I think LSJL does more than just pure Hydraulic stimulation does.

“Osteoblasts [orginiate] from MSCs”

“Our group previously presented a longitudinal study of bone marrow MSC quantification under DHS in a rat HLS model, where the MSC number was greatly increased in response to DHS by day 14 and diminished by day 21. MSCs may have completed proliferation and begun to differentiate towards osteoblastogenesis at this stage. In the meantime, the induced mRNA levels of the selected genes that we observed in the present study may couple the process of transforming MSC proliferation and differentiation into osteoblasts, which commit to the subsequent bone formation.”<-so maybe why the pro-chondrogenic genes were low at day 7 is that MSCs were proliferating rather than differentiating into chondrocytes.

Using chondrocyte hypertrophy to grow taller via articular chondrocytes?

It’s been established that the fingers can grow longer after cessation of development.  One primary difference of the fingers and the bones of say the legs is that there’s periosteum covering the articular cartilage of the legs and not of the bones of the fingers.  One issue of articular chondrocyte hypertrophy is it’s correlation with osteoarthritis(and).

Chondrocyte hypertrophy in skeletal development, growth, and disease.

” Chondrocyte hypertrophy is an essential contributor to longitudinal bone growth, but recent data suggest that these cells also play fundamental roles in signaling to other skeletal cells, thus coordinating endochondral ossification. On the other hand, ectopic hypertrophy of articular chondrocytes has been implicated in the pathogenesis of osteoarthritis.”

“Chondrocytes first differentiate from mesenchymal precursor cells after these have undergone cellular condensation{we have to be sure we’re covering this step if we want to create new growth plates}. This process, termed chondrogenesis, is characterized by expression of the cartilage master transcription factor Sox9 and induction of its target genes, for example, the classical chondrocyte markers, collagen II, and aggrecan. In a subsequent step, chondrocytes increase their rate of proliferation in a directed pattern along the developing longitudinal axis of the future bone, forming characteristic columns of clonal cells. Under tight control from endogenous and exogenous factors, these cells then withdraw from the cell cycle and start differentiating further. This differentiation step is accompanied by a large increase in cell volume, for example, chondrocyte hypertrophy. The conventional view is that hypertrophic chondrocytes represent the terminal step in this maturation process which culminates in the apoptosis of cells and replacement of hypertrophic cartilage by bone tissue. However, there have been reports about “transdifferentiation” of hypertrophic chondrocytes to osteoblasts”

“Not only is the cell volume increase during hypertrophy a major contributor to longitudinal bone growth, but these cells also act as signaling centers that secrete growth factors, cytokines, and other signaling molecules that can act on other cell types involved in endochondral ossification, such as osteoclasts, osteoblasts, and endothelial cells. Thus, the differentiation to this stage, as well as the behavior of differentiated hypertrophic chondrocytes, are tightly regulated by a multitude of systemic and local factors, including growth hormone, insulin-like growth factors (IGFs), thyroid hormone, parathyroid hormone-related peptide (PTHrP), Indian hedgehog, fibroblast growth factors (FGFs), canonical and noncanonical Wnt signaling, transforming growth factor-β (TGFβ) family members, C-type natriuretic peptide, and others. Within the cell, the classical mediators of these signals, such as β-catenin in canonical Wnt signaling and Smad proteins in TGFβ/bone morphogenetic protein signaling control cartilage growth, along with common kinases (e.g., MAP and PI3K/Akt) and GTPases (e.g., Rho GTPases). In the cell nucleus, Runx and MEF2 transcription factors, along with the histone deacetylase HDAC4, have been recognized as key regulators of chondrocyte hypertrophy.”

“mice lacking both FoxA2 and FoxA3 activity in cartilage display postnatal dwarfism, a result of severely impaired chondrocyte hypertrophy and mineralization shown by markedly attenuated expression of hypertrophic markers, such as collagen X, MMP13, and alkaline phosphatase”

“induction of chondrogenesis in chicken limb-bud mesenchymal micromass cultures results in increased FoxA1 and FoxA2 expression. Electrophoretic mobility shift assays show FoxA2 to bind to conserved binding sites within the chicken collagen X enhancer. Interestingly, exogenous FoxA2 can activate expression of a Collagen X-luciferase reporter gene in both chondrocytes and fibroblasts”

“HIF-2α contributes to physiological endochondral ossification by controlling chondrocyte hypertrophy, cartilage degradation, and vascularization. In particular, HIF-2α was identified as the most potent activator of the COL10A1 promoter in vitro. The expression of HIF-2α’s encoding gene, Epas1, also increased in parallel to that of Col10a1, Mmp13, and Vegf-α during chondrocyte differentiation”

“ectopic expression of Epas1 enhances cytokine-induced expression of catabolic genes necessary for cartilage breakdown. Similarly, both in vivo and in vitro models of Epas1 deficiency show protection against OA through suppression of cartilage degradation and catabolic and hypertrophic gene expression”

” HIF-2α [is a] functional inducer of the CEBPB promoter. As C/EBPβ is a transcription factor crucial for the transition from proliferation to hypertrophy in chondrocytes ”

“HDAC4 (histone deacetylase 4), which inhibits hypertrophy by suppressing the activity of Runx2 and MEF2C”

“kinase SIK3 (salt-inducible kinase 3) is essential to locate HDAC4 in the cytoplasm (and thereby relieve the inhibitory activity of HDAC4 on MEF2C and possibly Runx2). Correspondingly, chondrocytes from Sik3 KO mice show increased nuclear staining for HDAC4, resulting in delayed hypertrophy and dwarfism”

” Hdac3 KO mice showed accelerated chondrocyte hypertrophy but smaller cell size, which the authors attribute to increased expression of the phosphatase leucine-rich repeat phosphatase 1 (Phlpp1) in HDAC3-deficient chondrocytes. Increased Phlpp1 levels then suppress Akt signaling. Since the PI3K-Akt pathway is a major positive regulator of chondrocyte hypertrophy, these data suggest a direct pathway from epigenetic regulation of gene expression to cell size increase”

“Three different phases of hypertrophic cell enlargement, the first characterized by simultaneous increase in cell volume and dry mass, the second by preferential cell swelling without corresponding dry mass production, and the third again by proportional increases in cell volume and dry mass. Differences between slow and fast growing bones were due to changes in phase II and particularly phase III”

mice deficient for the key collagenase in OA, MMP13, still show chondrocyte hypertrophy in response to OA-inducing joint surgery, but are protected from cartilage degeneration“<-So it’s possible to grow taller via articular cartilage growth without cartilage degeneration.

We know that cartilage hypertrophy plays a very large role in how much bones grow longer.  There are supplements that could stimulate chondrocyte hypertrophy but inhibit cartilage degradation.  So this sort of method would be possible to test with suppelements only and not need mechanical methods. Cartilage degradation is vital though for longitudinal bone growth via growth plates so this would only work via adults past the developmental stage.

The chondrocytic journey in endochondral bone growth and skeletal dysplasia.

“Extracellular signals, including bone morphogenetic proteins, wingless-related MMTV integration site (WNT), fibroblast growth factor, Indian hedgehog, and parathyroid hormone-related peptide, are all indispensable for growth plate chondrocytes to align and organize into the appropriate columnar architecture and controls their maturation and transition to hypertrophy. Chondrocyte hypertrophy, marked by dramatic volume increase in phases, is controlled by transcription factors SOX9, Runt-related transcription factor, and FOXA2.”

“In vertebrates, the endochondral bones of the axial and appendicular skeleton develop from mesenchymal progenitors that form condensations of bipotential osteo-chondroprogenitors in the approximate shape of the future skeletal elements. These osteo-chondroprogenitors undergo lineage restriction toward either chondrocytes or osteoblasts. The primordial cartilage continues growing through recruitment of more mesenchymal progenitors and proliferation of chondrocytes. Once committed to the chondrocytic fate, the chondroprogenitors further differentiate into chondrocytes which proliferate, mature, exit the cell cycle, and undergo hypertrophy, forming an avascular cartilaginous template surrounded by a perichondrium. Mature hypertrophic chondrocytes secrete matrix vesicles which mediate cartilage calcification. At the same time, adjacent perichondrial cells differentiate into osteoblasts and secrete bone matrix, forming a bone collar surrounding the hypertrophic chondrocytes, then start to organize a periosteum and later become the shaft (diaphysis) of the bone. The primary ossification center begins to develop, with blood vessels penetrating the periosteum and gaining access to the calcified cartilage, bringing in osteoclasts/chondroclasts to degrade the cartilage. Subsequently, the osteoblasts lay down bone matrix to form trabecular bone. Gradually, the chondro-osseous junction forms, dividing the cartilage template into two ends, called epiphyses. These progressive steps of chondrocyte differentiation lead to the formation of a specialized structure called the growth plate ”

“The round chondrocytes of the RZ serve as a pool of progenitor-like cells for subsequent proliferation and differentiation. In the PZ, the round cells divide, become flattened, and organize into columns in the direction of longitudinal growth while proliferating. In the PHZ, the chondrocytes exit the cell cycle and initiate hypertrophic differentiation, characterized by the expression of Ihh (encoding the paracrine factor Indian hedgehog) and Col10a1 (encoding collagen type X). In the HZ, the size of the chondrocytes increases dramatically in the upper hypertrophic zone, and then these hypertrophic chondrocytes terminally differentiate in the lower hypertrophic zone, where the cartilage becomes calcified and is replaced with bone. The fate of hypertrophic chondrocytes at the chondro-osseous junction is controversial, with both cell death and survival being indicated”

Absence of Sox9, BMP2, BMP4, Smad4, Hif1a, and hoxd13 in MSCs all result in reduced height.  In proliferating chondrocytes loss of IHH+Gli3, Wnt5a, Wnt5b, Smad1, Smad5, Col2a1, and Acan result in reduced height.  In hypertrophic chondrocytes loss of Runx2 and Col10a1 results in reduced height.

“after cell aggregation and cluster formation, BMP signaling promotes cell–cell association through compaction of the aggregate.”

” the growth plates of different bones within the same animal grow at different rates. In part, the difference may be due to the differential duration of the G1 phase in proliferating chondrocytes, suggesting that cell cycle genes regulating G1 progression are of special importance in regulating endochondral bone growth. Progression through the different phases of the cell cycle is controlled by cyclin-dependent kinases (CDKs). The activities of CDKs are in turn regulated in many ways: (1) through the concentrations of their partner proteins, the cyclins; (2) through the concentrations of inhibitory proteins of the CIP/KIP and INK families (CDK inhibitors or CKIs); and (3) through inhibitory and stimulatory phosphorylation of various CDK residues. The principal targets of the CDKs are the pocket proteins RB, RBL1 (also known as p107), and RBL2 (also known as p130). Hypophosphorylated pocket proteins form complexes with transcription factors of the E2F family. Active CDKs phosphorylate the pocket proteins, causing them to dissociate from E2F, freeing the latter to activate target genes involved in cell-cycle progression and DNA replication. Cyclins D1, D2, and D3 are implicated in chondrocyte proliferation. The Cyclin D1 gene is a target of several paracrine factors, including parathyroid hormone-related peptide (PTHrP), IHH, and WNT5b. Mice lacking only cyclin D1 are viable but small, with a smaller PZ in the growth plate. Mouse embryos lacking all three cyclin Ds survive until about E13.5—15.5, suggesting that redundancy exists between the cyclin Ds in proliferation, and that they are regulated by different factors. RB, p107 and p130, regulate cell cycle exit and differentiation of growth plate chondrocytes. The CKI p57Kip2 is strongly expressed in terminally differentiated cells and can mediate cell cycle exit. Mice lacking this gene have short limbs and display endochondral ossification defects, as illustrated by delayed cell cycle exit and incomplete differentiation of hypertrophic chondrocytes. PTHrP and insulin-like growth factor-2 (IGF2) are reported to suppress p57Kip2 gene expression to mediate proliferation. Both IGF2 and CDKN1C (p57Kip2) are located in an imprinted locus and associated with Beckwith-Wiedemann syndrome, an overgrowth syndrome in human.”

“IHH promotes the differentiation of round proliferative chondrocytes in the resting zone into flat column-forming chondrocytes in a Gli3-dependent manner in mice”

“While IHH promotes proliferation, PTHrP delays exit from the cell cycle and initiation of hypertrophic differentiation. PTHrP represses the expression of the cell cycle inhibitor p57Kip2, and promotes the expression of the transcriptional coregulator ZFP521 and cyclin D1 to maintain chondrocyte proliferation”

“PP2A, when activated by PTHrP, dephosphorylates HDAC4, which is then translocated into the nucleus, where it inhibits the function of RUNX2 and MEF2C, and thus inhibits chondrocyte hypertrophy. Conversely, SIK3 can bind to HDAC4 and anchor it in the cytoplasm, leaving MEF2C and RUNX2 free to activate downstream targets to promote chondrocyte hypertrophy”

“Borderline chondrocytes adjacent to the bone collar may differentiate into osteoblasts{It is good for our purposes if chondrocytes transdifferentiate into osteoblasts because that means that they retain some of the chondrocyte genetic coding}. In chick explant culture, surviving hypertrophic chondrocytes undergo asymmetric cell division: one behaves as an osteoblast and may undergo further division while the other undergoes programmed cell death, leaving cell debris in the lacuna. The newly formed osteoblasts in the cartilage lacuna will be released into the chondro-osseous junction and contribute to endochondral bone formation .  Some of the hypertrophic chondrocytes look darker under electron microscopy. They are referred to as dark chondrocytes and are proposed to undergo programmed cell death inside the cartilage lacuna”

“[Hypertrophic chondrocytes] have been shown to undergo osteoblastic differentiation in situ when Sox9 is eliminated from prehypertrophic chondrocytes, displaying up-regulation of Runx2, Osx, Alpl, and Col1a1”

“In vitro, isolated chick hypertrophic chondrocytes can differentiate into osteoblasts in the presence of retinoic acid”

“[H1fa] induces collagen prolyl 4-hydroxylase expression in chondrocytes, which is necessary for generating 4-hydroxyprolines for the stability of newly synthesized collagen molecules”

What about avoiding dedifferentiation?

Spontaneous differentiating primary chondrocytic tissue culture: a model for endochondral ossification.

Primary cartilage-derived cell cultures tend to undergo dedifferentiation, acquire fibroblastic features, and lose most of the characteristics of mature chondrocytes{We want the opposite for it to undergo endochondral ossification, this dedifferentiation could be due to lack of mechanical stimuli}. This phenomenon is due mainly to the close matrix-cell interrelationship typical of cartilage tissue, which is vital for the preservation of the cartilaginous features. In this study we present a model for spontaneous redifferentiation of primary chondrocytic culture. Mandibular condyles excised from 3-day-old mice, thoroughly cleaned of all soft tissue, were digested with 0.1% collagenase. These mandibular condyle-derived chondrocytes (MCDC) were cultured under chondrogenesis-supporting conditions; that is, 5 x 10(5) cells/mL were incubated in Dulbecco’s modified Eagle medium supplemented with 100 microg/mL ascorbic acid, 1 mmol/L calcium chloride, 10 mmol/L beta-glycerophosphate, 10% fetal calf serum, and antibiotics. Development and growth rates of these cartilage-derived cultures were determined by following morphological and functional changes. MCDC proliferated intensively during the first 24-48 h following plating, showing fibroblast-like (long spindle-shaped) morphology and producing mainly type I collagen. The proliferation rate gradually declined, and the cells developed polygonal shapes and started to produce type II collagen. In the 10-14-day-old cultures, cells began to aggregate in cartilaginous nodules and exhibited positive staining for acidic Alcian blue, type X collagen, and von Kossa. Expression of core-binding factor alpha(1) increased between 3 and 5 days and declined gradually thereafter. The condylar-derived tissue culture presented here depicts a spontaneous redifferentiation chondrocytic tissue culture that exhibits features of mature chondrocytes typically found in skeletal growth centers. The present study offers a model for primary chondrocytic tissue culture, which might serve as a model for in vitro endochondral ossification.”

” chondrocytes of the mandibular condyle and the epiphyseal growth plate (EGP) are similarly regulated under both physiological and pathological conditions. Condylar chondrocytes express receptors for growth hormone, IGF-1, and parathyroid hormone and react similarly to the EGP chondocytes in type I diabetes and metabolic acidosis.”

New study provides more evidence that Lithium could have height growth applications

Lithium inhibits GSK-3Beta and this next study provides evidence that inhibition of GSK-3Beta can enhance height growth.

Inactivation of glycogen synthase kinase-3β up-regulates β-catenin and promotes chondrogenesis.zhou2014

“[Does] inhibition of glycogen synthase kinase-3β (GSK-3β) promote chondrocytes proliferation? The expression pattern of GSK-3β was firstly determined by immunohistochemistry (IHC) in normal mouse. Tibias were then isolated and cultured for 6 days. The tibias were treated with dimethylsulfoxide (control) or GSK-3 inhibitor SB415286 (SB86). Length of tibias was measured until 6 days after treatment. These bones were either stained with alcian blue/alizarin red or analyzed by IHC. In addition, GSK-3β and β-catenin were analyzed by Western blot. Finally, cartilage-specific GSK-3β deletion mice (KO) were generated. Efficiency of GSK-3β deletion was determined through Western blot and IHC. After treated by inhibitor SB86[the GSK-3B inhibitor], the overall length of growth plate was not changed. However, growth of tibia in SB86 group was increased by 31 %, the length of resting and proliferating was increased 13 %, whereas the length of hypertrophic was decreased by 57 %. Besides, the mineralized length was found to be significant longer than the control group. In KO mice, growth plate and calvaria tissue both exhibit significant reduction of GSK-3β whereas the lengths of tibias in KO were almost same compared with control mice. Finally, an increase amount of β-catenin protein was observed in SB86 (P < 0.05). In addition, significantly increased β-catenin was also found in the growth plate of KO mice (P < 0.05). Inhibition of GSK-3 could promote longitudinal growth of bone through increasing bone formation. Besides, the inactivation of GSK-3β could lead to enhancing β-catenin, therefore promote chondrocytes proliferation.”

It’s important to remember that growth rate does not always equal height attained due to natural development.  They also note that GSK-3B cartilage specific knock-out mice have almost the same tibia length as the control mice whereas mice that takes the GSK-3Beta inhibitor have longer tibias.  This is much more promising in regards to a supplement enhancing longitudinal bone growth as it’s much easier to ingest a GSK-3Beta inhibitor than to alter the genes on a molecular level.

“b-catenin is essential in determining whether mesenchymal progenitors will become osteoblasts or chondrocytes”

“chondrocytes may be influenced by GSK3b through Wnt/b-catenin signaling”

If you look at figure 3 you can see how great an increase in bone length it is.

Mechanical loading’s affect on stem cells

Previously, we learned that despite the demand for stem cells, the body did not produce more stem cells to complicate and demand greater than supply could lead to cancerous changes in the body.

Mechanical strain downregulates C/EBPβ in MSC and decreases endoplasmic reticulum stress.

“Exercise prevents marrow mesenchymal stem cell (MSC) adipogenesis, reversing trends that accompany aging and osteoporosis. Mechanical input, the in-vitro analogue to exercise, limits PPARγ expression and adipogenesis in MSC. We considered whether C/EBPβ might be mechanoresponsive as it is upstream to PPARγ, and also is known to upregulate endoplasmic reticulum (ER) stress. MSC (C3H10T1/2 pluripotent cells as well as mouse marrow-derived MSC) were cultured in adipogenic media and a daily mechanical strain regimen was applied. We demonstrate herein that mechanical strain represses C/EBPβ mRNA (0.6-fold ±0.07,) and protein (0.4-fold ±0.1) in MSC. SiRNA silencing of β-catenin prevented mechanical repression of C/EBPβ. C/EBPβ overexpression did not override strain’s inhibition of adipogenesis, which suggests that mechanical control of C/EBPβ is not the primary site at which adipogenesis is regulated. Mechanical inhibition of C/EBPβ, however, might be critical for further processes that regulate MSC health. Indeed, overexpression of C/EBPβ in MSC induced ER stress evidenced by a dose-dependent increase in the pro-apoptotic CHOP (protein 4-fold ±0.5) and a threshold reduction in the chaperone BiP (protein 0.6-fold ±0.1; mRNA 0.3-fold ±0.1).  ChIP-seq demonstrated a significant association between C/EBPβ and both CHOP and BiP genes. The strain regimen, in addition to decreasing C/EBPβ mRNA (0.5-fold ±0.09), expanded ER capacity as measured by an increase in BiP mRNA (2-fold ±0.2,) and protein. Finally, ER stress induced by tunicamycin was ameliorated by mechanical strain as demonstrated by decreased C/EBPβ, increased BiP and decreased CHOP protein expression. Thus, C/EBPβ is a mechanically responsive transcription factor and its repression should counter increases in marrow fat as well as improve skeletal resistance to ER stress.”

“The positive effect of exercise on the skeleton depends, at least partially, on the ability of mechanical input to regulate output of osteoblasts from progenitor mesenchymal stem cells (MSC){and the ability to regulate output of chondrocytes from progenitor mesenchymal stem cells}. Decreased adipocytes and increased pre-osteoblasts have been demonstrated in the marrow of running rats ”

“mechanical input applied to MSC slows adipogenesis in a process marked by downregulation of PPARγ as well as activation of β-catenin “

Breakthrough: Another study shows mechanical loads can alter height growth

Previously, I wrote another study about how mechanical loading can shape and alter joint and growth plate development.

If mechanical loading can alter limb development, then not only can mechanical loading regimes be used to increase height during development but also possible after growth plate fusion by creating new growth plates.  In fact one of the genes associated with the pre-growth plate cells in the zone of Ranvier is the mechanically sensitive to activation CMF608.

Mechanoadaptation of developing limbs: shaking a leg.

“The developing skeleton experiences mechanical loading as a result of embryonic muscle contraction. Embryos [may] coordinate the appearance of skeletal design with their expanding range of movements. Embryo movement [has a large role] in normal skeletal development; stage-specific in ovo immobilisation of embryonic chicks results in joint contractures and a reduction in longitudinal bone growth in the limbs. Epigenetic mechanisms allow for selective activation of genes in response to environmental signals, resulting in the production of phenotypic complexity in morphogenesis; mechanical loading of bone during movement appears to be one such signal. It may be that ‘mechanosensitive’ genes under regulation of mechanical input adjust proportionality along the bone’s proximo-distal axis, introducing a level of phenotypic plasticity{in other words it’s possible to alter how tall you will grow via mechanical factors}. If this hypothesis is upheld, species with more elongated distal limb elements will have a greater dependence on mechanical input for the differences in their growth, and mechanosensitive bone growth in the embryo may have evolved as an additional source of phenotypic diversity during skeletal development.”

“Cell movement-generated forces influence condensation of cartilage elements in developing limbs. There is also evidence that fundamental processes, including growth, differentiation, death and directional motility of cells, are likely guided by forces exerted by the cell cytoskeleton. This conforms with ‘tensegrity’ principles, with differential growth patterns producing local extracellular matrix distortion and the generation of tension in the cytoskeleton of associated cells.”<-LSJL would induce local extracellular matrix distortion and generate tension in the cytoskeleton of cells.

“dynamic loading in adult bones produces extracellular fluid flow within the bone’s lacunar-cannalicular system, which is detected by osteocytes”<-The idea is that LSJL goes further to induce MSCs to differentiate into growth plate plate chondrocytes.

“Embryonic muscle contraction appears to be necessary for the formation of bone ridges, which act as anchoring points for muscle attachment and are therefore important in the transduction of muscle-induced loading via tendons to the skeleton.”

“immobilisation of embryonic chicks alters cellular organisation of the interzone and results in changes in shape of the distal femur and proximal epiphysis of the tibiotarsus and fibula. After cavitation occurs, maintenance of joint cavities is also dependent on mechanical input. Post-cavitation induction of flaccid paralysis with pancuronium bromide, a non-depolarising neuromuscular blocker, also leads to loss of the joint cavities. Rigid paralysis induced with DMB, a depolarising neuromuscular blocking agent, causes muscle contraction and has been shown to partially maintain joint cavities “<-But there is there just a threshold of mechanical loading that is needed for proper development or can we enhance this development with enhanced mechanical loading.

“Detailed ‘targeting’ of specific temporal windows during development indicates that the effects of in ovo paralysis on bone length become significant at approximately E13 of development. This indicates that embryo bone growth is initially not sensitive to mechanical stimulus, but that mechanosensitivity is acquired later during development. This suggests that intrinsically regulated initial limb growth ‘switches’ later to regulation dominated by extrinsic factors such as mechanical signals. It remains to be determined whether this immobilisation-related skeletal growth retardation is due to deficient chondrocyte proliferation, differentiation, matrix synthesis or hypertrophy or due to insufficient replacement of calcified cartilage by bone during the endochondral ossification process. It has been suggested that mechanical loading regulates the elongation of chondrocyte columns during zebrafish craniofacial development”

evidence for mechanosensitivity in skeletal development is provided by observations of increased limb bone length when the level of embryo motility is increased in chicks. Incubation temperature increases embryo movement, with a 1 °C increase in incubation temperature producing a significant increase in embryo motility. This is associated with an increase in the number of myonuclei in embryo limb muscles and increased limb element lengths“<-Now this is the kind of fact we’re looking for.  So people can make their kids taller but what about us.  But we’d have to find the equilibrium temperature.

“This increase in limb length with temperature did not become significant until E12.5, providing further evidence that mechanosensitivity in skeletal element growth is acquired at a relatively late stage of development. Treatment with 4-aminopyridine (4-AP), a drug which stimulates the release of acetylcholine, thereby increasing its availability at the synaptic cleft and resulting in skeletal muscle hyperactivity, also stimulates embryo movement. Increases in tibia and femur lengths have been reported in chick embryos treated with 4-AP at E15 and E16, but not E14 ”

” The expression of IHH and hypertrophic markers such as MMP13 have been shown to be regulated in chondrocytes in vitro by cyclic mechanical stress”<-Although these genes wouldn’t be able to form new growth plates except possible IHH.  Mesenchymal Stem Cells transfected by IHH were induced to become chondrocytes in one study.

“In ovo immobilisation has been shown to alter expression patterns of COL X and IHH in embryonic limbs, suggesting that these genes are involved in linking mechanical stimuli from embryonic muscle contraction with regulation of bone formation in the limbs”

Unfortunately, this study only shows examples where longitudinal bone growth was altered in a very small window during embryonic development.  But increases the amount of evidence provided that mechanical loading can alter longitudinal bone growth which will eventually lead to prove that a specific mechanical loading regime such as that of LSJL may induce mesenchymal stem cells to become growth plate chondrocyte pre-cursors and form micro-growth plates.

Study shows feasability of micro-growth plates

The idea of LSJL is to create microgrowth plates via fluid shear strain on the mesenchymal stem cells in the bone marrow.  This study shows that microgrowth plates can exist:

Growth Plate Regeneration Using Polymer-Based Scaffolds Releasing Growth Factor

“Depending on the type of growth plate fracture and the severity it can lead to stunted bone growth or bone growth deformation. The current treatment options for growth plate fracture are removal of the bony bar and replacing it with a filler substance, such as, bone cement or fat, but still yield poor results 60% of the time. In previous work, poly(lactic-co-glycolic acid) (PLGA) scaffolds were developed and studied in vivo for the purpose of growth plate regenerationDepending on the type of growth plate fracture and the severity it can lead to stunted bone growth or bone growth deformation. The current treatment options for growth plate fracture are removal of the bony bar and replacing it with a filler substance, such as, bone cement or fat, but still yield poor results 60% of the time. In previous work, poly(lactic-co-glycolic acid) (PLGA) scaffolds were developed and studied in vivo for the purpose of growth plate regeneration”

micro growth platemicrogrowthplate

“Figure 6.7. Fat implant showed thin, continual line of cells across medial side that contained reserve (R), proliferative (P), hypertrophic (H) cartilage cells and calcification zones (C).”

Another micro growth plate:

microgrowth plate2

“Blank scaffold on (A) the lateral side with columnar structure and (B) the medial side with the appearance of stacked (S), reserve (R), proliferative (P), and hypertrophic (H) cartilage cells.”

Here’s a growth plate but loaded with IGF-1 so it’s much more sophisticated:

igf1 microgrowthplate

“IGF-I loaded scaffold showed dispersed pockets of cartilage cells throughout the medial side with the appearance of reserve (R), proliferative (P), hypertrophic (H), and degenerative zones (D).”

“In this study, the attempt to regenerate the growth plate did not result in columnar structure to the degree that the native growth plate has, regardless of the treatment type. It appeared that the fat implant allowed for some cartilage regeneration, but it was only a cell wide at most points and most of the chondrocytes were in the calcification zone. The tissue surrounding the cartilage areas was woven bone, which has been known to appear after fractures{But would this still result in a longer bone?}. The blank scaffold treatment resulted in tissue having a similar structure to that for the fat implants with a couple exceptions. First, there were a few areas where blank scaffolds had been placed with some cellular stacking, and secondly, the lateral side retained more structure, resembling that of the native growth plate, compared to defects treated with fat graft. The blank scaffolds gave the epiphyseal region more structural support, preventing further collapse of the lateral growth plate, while the fat graft implant had a thinner growth plate region across the whole tibia.”

“The defects treated with IGF-I-loaded scaffolds, both with or without seeded cells, showed a similar appearance on the lateral side as that of the blank scaffold group, however the medial sides were quite different. Without cells, the IGF-I-loaded scaffold resulted in pockets of chondrocytes throughout the medial side along the epiphyseal line that contained cells in all zones of cartilage development. The addition of cells created a large vertical pocket (~3 mm long) of chondrocytes located in the upper epiphyseal region. Interpretation of the IGF-I loaded samples was limited because only one sample could be used for observation so it is difficult to say if this cellular organization would occur again. The cells were mostly in the hypertrophic state and had no columnar organization. Both types of IGF-I loaded scaffolds (with and without cells seeding) increased the density of hypertrophic chondrocytes compared to the fat, blank, and hybrid scaffolds. Cells seeded on scaffolds containing IGF-I created the largest population of chondrocytes”

“Though the results did not show total growth plate regeneration, the necessary cell types were present”

It should be noted the mesenchymal stem cells used in this study were harvested from the diaphysis thus providing evidence that MSCs needed to create growth plates do not necessarily have to be from the Zone of Ranvier.

The study did not display changes in length due to the various scaffolds.  The fat scaffold and IGF-1 seeded scaffold did reduce the angular measurement resulting from part of the growth plate being damaged.  The blank scaffold altered the angular measurement disparity but increased it in the tibia and decreased it in the femur.  We can be fairly certain that microgrowth plates can alter longitudinal bone growth as the angular measurement is dependent on how tall one side of the bone grows versus the other.

Even though this study involves scaffolds and LSJL does not.  The information about microgrowth plates altering height growth can be extrapolated to LSJL as MSCs could migrate to the epiphyseal region and use bone as a natural scaffold.

This study provides evidence that you don’t need to create a whole growth plate to increase height.